AU2007263265A1 - Differential IL-4 and/or IL-10 cytokine expression in human cancer - Google Patents

Differential IL-4 and/or IL-10 cytokine expression in human cancer Download PDF

Info

Publication number
AU2007263265A1
AU2007263265A1 AU2007263265A AU2007263265A AU2007263265A1 AU 2007263265 A1 AU2007263265 A1 AU 2007263265A1 AU 2007263265 A AU2007263265 A AU 2007263265A AU 2007263265 A AU2007263265 A AU 2007263265A AU 2007263265 A1 AU2007263265 A1 AU 2007263265A1
Authority
AU
Australia
Prior art keywords
tumour
cytokine
apoptotic
cells
chemotherapeutic
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2007263265A
Inventor
Christian Gieffers
Oliver Hill
Giorgio Stassi
Meinolf Thiemann
Matilde Todaro
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Apogenix AG
Original Assignee
Apogenix AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Apogenix AG filed Critical Apogenix AG
Publication of AU2007263265A1 publication Critical patent/AU2007263265A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6863Cytokines, i.e. immune system proteins modifying a biological response such as cell growth proliferation or differentiation, e.g. TNF, CNF, GM-CSF, lymphotoxin, MIF or their receptors

Abstract

The invention concerns a method for diagnosing a cancer type, whereby the expression of anti-apoptotic cytokines is determined in the tumour cells. The differential diagnosis of the present invention is used to classify tumour disorders and to recommend the required treatment and to monitor the progress and response to the treatment.

Description

WO 2007/147600 PCT/EP2007/005480 Differential cytokine expression in human cancer Description 5 The invention concerns a method for diagnosing a cancer type, whereby the expression of anti-apoptotic cytokines in the tumour cells is determined. The differential diagnosis of the present invention is used to classify tumour disorders and to recommend the required treatment and to monitor the 10 progress and response to the treatment. The balance between cell survival and cell death is controlled by pro apoptotic and anti-apoptotic factors, whose dysregulation contributes to the development of several pathological conditions, including cancer. High 15 expression of anti-apoptotic factors is commonly found in human cancers and contributes to both neoplastic cell expansion and resistance to the therapeutic action of cytotoxic drugs. It has already been reported that autocrine production of anti-apoptotic cytokines by tumour cells strongly modulates the susceptibility to the receptor and chemotherapy-induced 20 apoptosis. In particular, it has previously been reported that IL-4 and IL-10 act as autocrine growth factor in cancer cells inducing upregulation of anti apoptotic proteins, which protect the tumour cells from the death induced by chemotherapeutic drugs (Stassi et al., Cancer Res. 63, 6784-90 (2003), Todaro et al., Cancer Res. 66, 1491-9 (2006)). 25 Tumours are composed of a heterogeneous combination of cells, with different therapeutic characteristics and different proliferative potentials. In particular, cancer cells may give rise to phenotypically diverse progeny of cells, either endowed with a definite proliferative potential or having a limited 30 or no proliferative potential. In this respect recent evidence suggests that the tumourigenic growth capacity is in fact confined to a small subset of so-called cancer stem cells WO 2007/147600 PCT/EP2007/005480 2 (CSC). The International Application PCT/IT2005/000523 discloses a method for isolation and culturing of stem cells from solid tumours. This subpopulation of cancer cells can self-renew and give rise to a population of heterogeneous cells which exhibit diverse degrees of differentiation. 5 Moreover, it has recently been found that these cancer stem cells are significantly resistant to drug-induced apoptosis, thus escaping anti-tumour therapies and this being probably the underlying reason for chemotherapy inefficiency. 10 It has now been demonstrated that CSC predominantly produce IL-4 and IL 10 and are responsible for the above mentioned alteration of sensibility to drug-induced cell death. It has now also been found by the inventors of the present invention that 15 solid tumours may be differentiated in respect of anti-apoptotic cytokine expression level and/or profile. The expression of anti-apoptotic cytokines differs between individual tumours of the same organ and even within cells or portions of a single tumour. These results lead to new efficient strategies in the tumour diagnosis and/or therapy. 20 In particular, an object of the present invention was to provide a method which allows the identification and diagnosis of cancer types and cancer cells which express anti-apoptotic cytokines. 25 Accordingly, the present invention provides a method for diagnosing tumour types, especially solid tumour types, using the anti-apoptotic cytokines as a target. Particularly, the invention refers to a method for diagnosing a cancer type comprising the steps of: (a) providing a sample from a solid tumour comprising tumour cells, 30 (b) determining the expression of at least one anti-apoptotic cytokine in said tumour cells, and (c) classifying the solid tumour as a non-cytokine expressing tumour or as a cytokine-expressing tumour.
WO 2007/147600 PCT/EP2007/005480 3 Hence, the invention concerns the differential diagnosis of cancer types by means of the determination and/or quantification of the expression profile and/or level of anti-apoptotic cytokines in the tumour sample. As anti 5 apoptotic cytokines, IL-4 and/or IL-10, particularly IL-4, is preferred. The differential diagnosis according to the invention allows to classify tumour types and to identify those which show expression of anti-apoptotic cytokines and which are refractory to treatment with chemotherapeutic 10 agents. Hence, the expression of anti-apoptotic cytokines is a significant marker for tumour classification which allows a selection of targeted therapeutic strategies. For example, the method of the invention may be useful to predict whether a 1s patient suffering from a certain cancer type would be resistant or susceptible to a certain therapy and to provide an optimised treatment strategy. According to the present invention, it was found that cancer types can be classified as non-cytokine-expressing tumours or as cytokine-expressing 20 tumours. When determining the expression of IL-4 and/or IL-10, more particularly, the expression of IL-4, the solid tumours may be classified with regard to their expression of either only IL-4 or only IL-10 or both IL-4 and IL-10. Therefore, 25 the method according to the present invention allows the differentiation between solid tumour classified as IL-4-expressing tumours or IL-4 non expressing tumours, solid tumour classified as IL-10-expressing tumours or IL-10 non-expressing tumours and solid tumour classified as IL-4 and IL-10 expressing tumours or non-IL-4 and non-IL10 expressing tumours. 30 The method of the present invention is preferably performed on solid tumours and in particular on epithelial tumours. Said epithelial tumours may be chosen from the group consisting of thyroid, breast, prostate, bladder, WO 2007/147600 PCT/EP2007/005480 4 colon, gastric, pancreas, kidney, liver and lung cancer. More preferably, the epithelial tumour is a colon, gastric, breast, lung, bladder or prostate cancer. The diagnostic method of the present invention may be performed on s various cell samples from a solid tumour. The test sample is preferably a cell sample from primary tumour and/or from the tumour environment isolated from a subject, e.g. a human patient. For example, tumour cell tissue obtained by biopsy, resection or other techniques can be tested. The tumour sample comprises tumour cells. The expression of anti-apoptotic cytokine in 10 the tumour cells is preferably determined on primary tumour cells and/or cancer stem cells. Methods for the determination of the anti-apoptotic cytokine expression in the tumour cells are well-known in the art. The determination of the 15 expression, in particular of the overexpression, of the anti-apoptotic cytokine in the tumour cell is conducted by the detection of said cytokine on the protein level and/or the nucleic acid level. The determination of cytokine proteins may be performed in the tumour cells 20 or in the tumour microenvironment. Methods to determine the presence and amount of cytokine proteins in a given sample are well known to the person skilled in the art and may be immunochemical methods such as immunohistochemistry, Western blotting, immunoprecipitation and ELISA methods. Further methods based on massspectrometry, comprising MALDI 25 MS, can be used to determine presence and amount of cytokine proteins. Cytokine nucleic acids are detected and quantified herein by any of means well known to those skilled in the art. Hybridization techniques together with optional amplification methods are frequently used for detecting nucleic 30 acids. Expression of cytokine mRNAs may for example be detected by Northern blot analysis or by reverse transcription and subsequent amplification by PCR.
WO 2007/147600 PCT/EP2007/005480 5 The method according to the invention may comprise the further step of (d) determining the sensitivity of the cells of a cytokine expressing tumour against at least one chemotherapeutic or pro-apoptotic agent in the presence and/or in the absence of an antagonist of said expressed 5 cytokine and/or its receptor. In order to investigate the sensitivity of the cytokine-expressing tumour cells to chemotherapeutic and/or pro-apoptotic agents, the viability of the tumour cells exposed to said chemotherapeutic or pro-apoptotic agents in the 10 absence and/or presence of cytokine neutralizing agents may be measured. Methods for determining the sensitivity of the tumour cells to a given agent are well known by those skilled in the art (e.g. as described in Examples). Based on this determination, the method according to the present invention 1s may further comprise the step of (e) selecting a cancer type-specific treatment. As already mentioned, the invention is based on the observation that solid tumours may be differentiated by their expression or degree of expression of 20 anti-apoptotic cytokines and in particular IL-4 and IL-10 cytokines. Since the expression of IL-4 and IL-10 anti-apoptotic cytokines in tumours or tumour cells is responsible for refractoriness to treatment, e.g. with chemotherapeutic and/or pro-apoptotic agents, the anti-apoptotic cytokines should be neutralized in order to increase the sensitivity of the tumour 25 towards treatment. Thus, the invention may also encompass an examination of the sensitivity or resistance to chemotherapeutic and/or pro-apoptotic agents in combination with antagonists of a cytokine expressed by the tumour. 30 In a preferred embodiment, the sensitivity assay performed in step (d) of the method leads to the determination of a chemotherapeutic or a pro-apoptotic agent against which the cell of the cytokine-expressing tumours are particularly sensitive.
WO 2007/147600 PCT/EP2007/005480 6 Consequently, according to step (e) of the present invention, a successful tumour type-specific treatment may be selected comprising the administration of a combination of a cytokine-neutralizing agent and a 5 chemotherapeutic or pro-apoptotic agent. A cytokine-neutralizing agent may be any compound which reduces the amount and/or activity of a cytokine. For example, the cytokine neutralizing agent may be an agent which inhibits a signal transduction pathway 10 triggered by the cytokine autocrinely expressed by the tumour cells. Hence, any agent is contemplated that is capable of modulating the expression and/or function of a cytokine directly and/or indirectly, namely affecting the expression and/or function of the respective cytokine protein and/or cytokine receptor. 15 Preferably, the cytokine neutralizing agent is an IL-4 and/or IL-10 neutralizing agent, i.e. any agent which is able to inhibit the signal transduction pathway triggered by the autocrine expression of IL-4 and/or IL 10. 20 Cytokine neutralizing agents may be selected, among others, from agents that inhibit and/or reduce the expressed cytokine protein activity, agents which degrade the expressed cytokine protein and agents that inhibit the cytokine production. Agents that block the cytokine activity are, for example, 25 antagonists which block the cytokine receptors, e.g. peptides, small molecules, muteine variants of the cytokines which show an antagonistic activity compared to the original signal of the cytokine. Examples for such muteins are in particular IL-4 muteins such as Aerolast* from Aerovance and Pitrakinra* and BAY-36-1677 from Bayer. Further antibodies against the 30 cytokine receptor or antibodies against the cytokine protein may be used. The antibody is preferably an antibody against IL-4 and/or IL-10, e.g. antibodies from Amgen and Immunex or an antibody against the IL-4 receptor and/or the IL-10 receptor, e.g. the antibody Pascolizumab* from WO 2007/147600 PCT/EP2007/005480 7 Glaxo. The antibody may be a complete antibody, e.g. an IgG antibody, or an antigen-binding fragment thereof. Preferably, the antibody is a monoclonal chimeric or humanized antibody which has human constant domains, e.g. human constant IgG1, IgG2, IgG3 or IgG4 domains. More 5 preferably, the antibody is a humanized antibody which additionally comprises human framework regions. Also preferred are antibody fragments, e.g. divalent or monovalent antibody fragments such as F(ab) 2 fragments. On the other hand, the antibody may be a recombinant antibody, e.g. a single chain antibody or a fragment thereof, e.g. an scFv fragment. 10 Soluble cytokine receptors, preferably without the membrane spanning and the intracellular domain, can also be used as agents blocking the cytokine activity. These soluble receptors are, for example, from Regeneron, in particular IL-4R/IL-13R-Fc fusion proteins, and soluble receptors from 15 Amgen and Immunex, in particular Nuvance* and Altrakincept*. Specific examples of soluble receptors comprise the extracellular domain (ECD) of a human IL-4 receptor, e.g. from a shortened ECD of human IL-4R alpha amino acid 24 to amino acid 224, 225, 226, 227, 228, 229 or 230 and optionally further domains, e.g. the extracellular domain of a human 11-13 20 receptor and/or a human Fc immunoglobulin domain. As preferred example of agents that degrade the expressed cytokine protein designer proteases can be mentioned in the context of the present invention. The production of the cytokine proteins can, on the other hand, be inhibited 25 for example by agents acting on the nucleic levels such as antisense nucleic acids, siRNA molecules and/or ribozymes. Preferred cytokine antagonists are described in the international patent application WO 2004/069274. Antibodies directed against cytokines are 30 preferably used as cytokine-neutralizing agents. Anti-IL-4 antibodies disclosed in European patent application EP-A-0 730 609 are especially suitable as cytokine-neutralizing agents of the method of the present invention. In a very preferred embodiment, the antibody derived from the WO 2007/147600 PCT/EP2007/005480 8 monoclonal antibody 6A1 produced by hybridoma cell line ACC93100620 or an antigen-binding fragment thereof is used as cytokine-neutralizing agent. The chemotherapeutic agent used in steps (d) and/or (e) is selected from 5 antimetabolites, DNA-fragmenting agents, DNA-cross-linking agents, intercalating agents, protein synthesis inhibitors, topoisomerase I and 11 inhibitors, micro-tubule-directed agents, kinase inhibitors, hormones and hormone antagonists. Particularly, the chemotherapeutic agent is selected from cisplatin, carboplatin and oxaliplatin. As preferred pro-apoptotic agents, 10 TRAIL and CD95 ligand can be selected. Based on the results obtained from the combined administration of anti therapeutic cytokine-antagonists and chemotherapeutic and/or pro-apoptotic agents to the tumour cell, a therapeutic strategy can be developed based on 1s a specific combination of drugs which has proven to be effective. A further object of the present invention is therefore the use of a combination of a cytokine-neutralizing agent and a chemotherapeutic or pro-apoptotic agent and the manufacture of a medicament for the tumour treatment, such 20 as a first line tumour treatment or as second or third line tumour treatment, e.g. for the treatment of refractory tumours, such as tumours which have become refractory against one or more anti-tumour agents. Thus, a further aspect of the present invention is the use of a combination of 25 (i) at least one cytokine-neutralizing agent and (ii) at least a chemotherapeutic or pro-apoptotic agent for the manufacture of a medicament for the treatment of a cancer type classified as cytokine-expressing tumour. 30 One of the main causes of drug resistance in tumour cells is based on the observation that a surviving small population of tumour cells, and in particular of tumour stem cells, after an apparently complete regression or surgical excision of the primary tumour could renew the tumour and WO 2007/147600 PCT/EP2007/005480 9 contribute to the so called minimal residual disease (MRD). In this respect, since the combination therapy is particularly suitable for increasing the therapeutic sensitivity of tumour stem cells, a further aspect of 5 the present invention is the use of a combination of (iii) at least one cytokine-neutralizing agent and (iv) at least a chemotherapeutic or pro-apoptotic agent for the manufacture of a medicament for the treatment of minimal residue disease. 10 According to a preferred embodiment of the present invention, the use of a combined therapy of the above agents (i) and (ii) can further be in combination with surgery and/or irradiation therapy. In particular, the medicament combination is for simultaneous, separate or sequential 15 combination therapy with surgery and/or irradiation therapy. According to one preferred embodiment of the present invention, the administration of agent (i) and agent (ii) is started simultaneously. Alternatively, the combination therapy can be started stepwise. According to 20 this preferred embodiment of the invention, the start of the administration of the cytokine-neutralizing agent (i) is 5 1 week before the administration of the chemotherapeutic or pro-apoptotic agent (ii). The administration of the chemotherapeutic or pro-apoptotic agent (ii) may in turn start 1 week before the administration of the cytokine-neutralizing agent (i). 25 Still a further embodiment of the invention is a soluble IL-4 receptor polypeptide or fusion polypeptide comprising a C-terminally shortened extracellular domain, e.g. a domain shortened by 1, 2, 3, 4, 5, 6, 7, 8 or more amino acids or a nucleic acid molecule encoding such a polypeptide. The 30 shortened extracellular domain may be derived e.g. from human IL-4 receptor alpha (NCBI accession NP 000409) which C-terminally ends at amino acid 230, 229, 228, 227, 226, 225 or 224. Preferably the C-terminal end is amino acid 224. The polypeptide may comprise at least one further WO 2007/147600 PCT/EP2007/005480 10 domain, e.g. an N-terminal signal peptide, a further effector domain, e.g. an IL-13 receptor extracellular domain, an Fc immunoglobulin domain, and/or a purification domain. An example of a shortened IL-4R polypeptide is described in Example 4. The shortened IL-4R polypeptide is suitable for 5 pharmaceutical applications, e.g. for the treatment of tumours, particularly for the treatment of IL-4-associated tumours as described above. The invention is further illustrated by the following examples: 10 Examples Materials and Methods Human Tissues. Cancer specimens were obtained at the time of surgical 15 treatment, in accordance with the ethical standards of the institutional commitee responsible for human experimentation. Whereas normal tissues were obtained from the controlateral part of the surgically removed tumour. Histological diagnosis was based on the behavioral microscopic features of carcinoma cells determining the histologic type and grade. 20 Human primary cell purification. Normal and cancer tissues were digested for 2 hours with collagenase (1.5 mg/ml) (Gibco BRL., Grand Island, NY) and hyaluronidase (20 pg/ml) (Sigma Chemical Co., St. Louis, MO) as previously described (1). Once digested, cells were maintained on plastic in DMEM 25 medium (EuroClone Ltd., West York, UK) at 37 0 C in a humidified atmosphere of 5% C02. Following 12 further hours of culture, cancer cells were allowed to grow in monolayer for the immunocytochemistry or detached with trypsin+EDTA for functional, protein expression and gene transcript levels analyses. For colon and gastric cells culture, plastic was 30 coated with/cm 2 of collagen (Calbiochem GmbH, Darmstadt, Germany). Cancer cells were cultured in presence or absence of human recombinant IL-4 (20 ng/ml), IL-10 (40 ng/ml) (Euroclone, Paignton, UK), neutralizing antibodies against human IL-4 (10 p/ml) (R&D Systems, Europe, Ud) for 48 WO 2007/147600 PCT/EP2007/005480 11 hrs. Anti-CD95 (mAb CH-1 1, IgM; Upstate Biotechnology Inc.) or control IgM (Sigma) or isoleucine zipper TRAIL (iz-TRAIL; 200 ng/ml) were used to determine sensitivity to CD95- or TRAIL-induced apoptosis in cancer cells. Moreover, following exposure to anti-IL-4 and anti-IL-10 cancer cells were 5 treated with oxaliplatin (100 pM) or doxorubicin (5 pM) or cisplatin (300 ng/ml), or taxol (5 pM) (Sigma) or etoposide (1 pM; Biomol, Plymouth Meeting, PA). Survival and death assays. To evaluate apoptotic events the DNA staining 10 and flow cytometry analysis were performed. The percentage of hypodiploid nuclei was evaluated as described in Stassi et al., Cancer Res. 2003, 63 (20):6784-90. Alternatively, human purified cancer cells were plated in 96 well plates in triplicate at 15,000 cells/well and cultured. The number of viable cells was detected by CellTiter Aqueous Assay Kit (Promega 15 Corporation, WI, USA) following the instructions of manufacturer. HuT78 cells plated at 2 x 150/mi and treated with CD95-activating antibody CH11 (200 ng/ml) were used as a positive control for cell death measurement. Immunohistochemical analysis. Immunohistochemistry was performed on 20 5 pm thick paraffin-embedded colon, gastric, prostate, breast, lung, liver, pancreas, kidney and bladder normal and tumour sample sections. Dewaxed sections were treated for 10 min in microwave oven in 0.1 M citrate buffer. Then, sections were incubated for 10 min with Tris Buffer Saline (TBS) containing 10% AB human serum to block the unspecific staining. After 25 elimination of excess serum, sections were exposed overnight at 40C to specific antibodies against IL-4 (B-S4 mouse IgG1, Caltag Laboratories, Burlingame, CA), IL-10 (B-N10 mouse IgG 2 ., Caltag), IL-4Ra (C-20 rabbit IgG Santa Cruz Biotechnology Inc, Santa Cruz, CA), IL-1 OR (C-20 rabbit igG Santa Cruz Biotechnology), TRAIL-R1 (HS101 mouse IgG1, Alexis 30 Biochemicals, Lausen, CH) TRAIL-R2 (HS201 mouse IgG1, Alexis) or isotype-matched controls at appropriate dilutions. Following exposure to primary antibody cells were treated with biotinylated anti-rabbit or anti mouse immunoglobulins, washed in TBS and then incubated with WO 2007/147600 PCT/EP2007/005480 12 streptavidin peroxidase (Dako LSAB 2 Kit, Dako Corporation Carpinteria CA, USA). Staining was detected using 3-amino-9-ethylcarbazole (AEC) as a colorimetric substrate. Counterstaining of cells was performed using aqueous hematoxylin. 5 RT-PCR analysis. Total RNA was prepared from cultured cells using the Rneasy Mini Kit (Qiagen GmbH, Germany) according to manufacturer's instructions. Reverse transcription and PCR amplification for each preparation with 1pg of total RNA was performed using OneStep RT-PCR 10 Kit (Qiagen). Two primers specific for the IL-4 coding sequence 5'-CCA CGG ACA CAA GTG CGA TA nucleotides 436-455 (exon 1) and 5'-CCT TGC AGA AGG TTT CCT TCT-3' complementary to nucleotides 564-584 (exon 3) (GenBank accession number NM 000589.2) were selected to specifically amplify IL-4. 15 GAPD gene was amplified from the same RNA preparations as housekeeping control (coding sequence 5'-TGA CAT CAA GAA GGT GGT GA-3' nucleotides 843-863 and 5'-TCC ACC ACC CTG TTG CTG TA-3' complementary to nucleotides 1033-1053; NM-002046 accession number). Thirty-five cycles were performed, each consisting of the following 20 conditions: 94"C, 30 sec; 58*C, 30 sec; 72"C, 30 sec. Protein isolation and western blotting analysis. Cell pellets were resuspended in ice-cold NP-40 lysis buffer (50 mM Tris-HCI, pH 7.5, 150 mM NaCl, 1 mM EGTA, 1% NP-40) containing protease inhibitors as 25 described in Stassi et al. Nature Immunology 2000, 1, 1-6. Immunoblotting of Abs specific for actin (Ab-1, mouse IgM, Calbiochem, Darmstadt, Germany), CD95L (G247-4, mouse IgG1, PharMingen, San Diego), CD95 (C-20, Santa Cruz Biotechnology), cFLIP (NF6 mouse IgG1, Alexis Biochemicals, Switzerland), PED/PEA-15 (rabbit IgG kindly provided by G. Condorelli), Bcl 30 2 (124, mouse IgG1, Upstate Biotechnology Inc.) and BcI-X (H-5, mouse IgG1, Santa Cruz Biotechnology) was detected by HRP-conjugated anti mouse or anti-rabbit Abs (Amersham Biosciences UK Limited, England) and visualized with the chemiluminescence detection system (SuperSignal West WO 2007/147600 PCT/EP2007/005480 13 Dura Extended duration Sustrate, Pierce, Illinois, USA). Example 1 5 Autocrine production of IL-4 in cancer cells In order to investigate if the tumour microenvironment influences cancer cell phenotype and function, the presence of IL-4 and IL-10 previously found to be autocrinely produced by cancer thyrocytes was evaluated. Immunohistochemistry analyses demonstrated that all the investigated solid 10 tumour histotypes expressed high levels of IL-4, while IL-10 was less detectable. Results are shown in Table 1. Table 1. Cytokine expression in cancer cells Cancer IL-4 IL-10 PTC ++++ +++ FTC ++++ +++ UTC ++++ ++++ Colon ++++ + Gastric +++++ - Lung ++++ + Pancreas + + Glioblastoma +++ ++ Prostate ++ + Breast ++++ + Bladder ++++ + Liver + + Kidney ++ ++ 15 Interestingly, the reactivity against IL-4 localized to colon, breast, lung, gastric, liver, prostate, pancreas, kidney and bladder cancer cells, suggesting that neoplastic cells are the source of high production for IL-4 and less for IL-10 (Table 1 and Fig. 1a). To exclude the possibility that the 20 reactivity observed in tumour cells was exclusively due to the release of type WO 2007/147600 PCT/EP2007/005480 14 2 cytokines by infiltrating T cells, freshly purified colon, breast, gastric and lung cancer cells were analyzed by RT-PCR. In agreement with immunohistochemistry results, IL-4 mRNA expression levels of purified cancer cells were highly increased compared to related normal cells (Fig. 1 5 b), demonstrating that autocrine production of IL-4 is not restricted to thyroid cancer cells but also takes place in other epithelial malignant cells from solid tumours which produce considerable amounts of IL-4. Epithelial cancer cells express high levels of anti-apoptotic proteins. 10 Colon, breast, gastric and lung cancer cells are resistant to death ligand and to chemotherapy-induced cell death. To determine the mechanism responsible for this refractoriness, it was investigated whether aberrant expression of anti-apoptotic factors could be implicated in the impaired "extrinsic" and "intrinsic" apoptotic signal pathway generated by death 1s ligands or chemotherapy. It was found by immunohistochemistry and Western blot analyses that epithelial carcinoma cells express CD95, TRAIL R1 and TRAIL-R2 (Fig. 2a and b). Therefore, the inventors of the present invention evaluated the presence and measured the expression levels of cFLIP, PED/PEA-15, Bcl-xL and Bcl-2 in colon, breast, gastric and lung 20 normal and cancer cells. While cFLIP and PED/PEA-15 levels were approximately three fold higher in freshly purified cancer cells, as compared with normal colon, breast and lung cells (Fig. 2a), Bcl-xL levels were four fold higher. Bcl-2 expression levels were only two fold higher in all the cancer cells analyzed, as compared with normal cells. Thus, anti-apoptotic genes 25 upregulation in colon, breast, gastric and lung cancer cells may confer resistance to CD95- TRAIL- and chemotherapy-induced apoptosis. IL-4 increases survival, growth of epithelial neoplastic cells. The expression of IL-4 receptor in both normal and neoplastic cells was 30 evaluated. Immunohistochemistry on paraffin embedded sections showed that IL-4 receptor was expressed in all the cancer tissues analysed. The results are shown in the following Table 2 and in Fig. 3a.
WO 2007/147600 PCT/EP2007/005480 15 Table 2. IL-4R expression in cancer cells Cancer IL-4R PTC ++++ FTC ++ UTC +++ Colon ++ Gastric +++ Lung +++++ Pancreas +++ Glioblastoma ++ Prostate ++ Breast +++ Bladder ++++ Liver +++ Kidney +++ 5 In order to investigate the possible involvement of IL-4 on tumour cell survival, colon, breast, gastric and lung normal cells were exposed to 20 ng/ml of IL-4 and analyzed for cell growth. IL-4 significantly increased the growth rate of colon, breast and lung normal cells (Fig. 3b). 10 Furthermore, to determine the involvement of IL-4 in the refractoriness of cancer cells to CD95, TRAIL and chemotherapeutic agents, normal colon, breast, gastric and lung cells were pre-incubated with IL-4 and then analyzed for expression of those anti-apoptotic proteins implicated in the death ligands and chemotherapy cell death resistance. IL-4 increased the 1s protein levels of cFLIP, PED/PEA-15, BcI-xL and Bcl-2 in normal colon, breast (Fig. 3c) and gastric and lung cells, suggesting that autocrine IL-4 production might protect cancer cells from chemotherapy and death receptor stimulation, up regulating anti-apoptotic factors. 20 WO 2007/147600 PCT/EP2007/005480 16 IL-4 neutralization promotes growth arrest and cell death induced by CD95, TRAIL and chemotherapy in cancer cells To directly demonstrate that autocrine production of IL-4 confers protection 5 from cell death induced by CD95, TRAIL and chemotherapy, we investigated the effects of IL-4 neutralization in colon, breast and lung cancer cells. Exposure of freshly purified colon, breast, gastric and lung cancer cells to neutralizing antibodies against IL-4 for 48 hrs sensitized cancer cells to chemotherapy- and death receptor-induced cell death confirming the anti 10 apoptotic role of IL-4 in solid cancer. The results are shown in the Figures 4a-c. Furthermore, IL-4 neutralization blocked colon, breast, gastric and lung tumour cell growth up to 15 days (Fig. 5) and down-modulated the protein 15 expression levels of cFLIP, PED/PEA-15, BcI-xL and Bcl-2. These data indicate that autocrine production of IL-4 might play an important role in growth control and is specifically required for survival of cancer cells. Tissue specimens from freshly operated tumour patients were screened for 20 IL-4 and IL-10 expression by a variety of standard methods such as RT PCR, western blots and immunohistochemistry. Likewise, the expression of their respective receptors was analysed by the same methods. Purified cancer cells were then tested for their sensitivity against chemotherapeutic agents such as e.g. etoposide, doxorubicin, oxaliplatin and apoptosis 25 inducers such as TRAIL and CD95 ligand. The results are shown in the following Table 3. 30 WO 2007/147600 PCT/EP2007/005480 17 Ifl 0 00 Cv, 0~ 6 m 04 0o '- V- c? to o Lo) cv, r-_ M C) 0 E V- V- 2 a) CL m C0 04 0 CD, 6% L cu 0 -. VII T- a) 0 co 2 2 Z c, E 04 - m~ c o0a E 00 U U) LO 0O 02 o 0 0 - 0 0 4. L& CD 4- c T e 0~ x ca a) O) U') co 14 CO _O C C 8 .ja) - CO 'm- V- Li) lir Z- =12 o~ CO co 1, Cf N .4 c C/) z CD .2 -W m E 2 * mf C 4 (A3 0 U) " 0.~ ~ _j 0D~ - 2 C CL -m~LU i. IL IL WO 2007/147600 PCT/EP2007/005480 18 As shown from the results in Table 3, it was surprisingly found that normally resistent primary tumour cells expressing IL-4 and/or IL-10 became sensitive against the tested chemotherapeutic agents and/or the pro-apoptotic agents 5 when incubated in the presence of an IL-4 antibody such that more than 90% of the cells died in a couple of days. Particular significant sensitisation to death-receptors and chemotherapy-induced cell death was shown for colon, gastric, breast, lung, prostate and bladder cancer cells. 10 Example 2 The data reported in this example reveal that purified colon cancer stem cells produce high levels of IL-4 and that the exposure of the cancer cells to neutralising antibodies against IL-4 sensitised cells to cytotoxic drug- and 15 TRAIL-induced apoptosis. Further, the following data show that a combined treatment of colon tumours with chemotherapeutic agents and anti-IL-4 agents significantly reduces tumour outgrowth. To investigate the sensitivity of colon CSC to chemotherapeutic drugs, the 20 viability of colon CSC spheroids exposed to cisplatinum (300ng/ml) and oxaliplatin (100 pM) was measured, doses equivalent to those reached during cancer treatment in vivo. In addition, colon CSC were treated with the apoptosis-inducing death ligand TRAIL (200 ng/ml). Primary (adherent) cells from human colon cancer specimens showed some sensitivity in vitro to all 25 three drugs tested, whereas colon CSC were significantly resistant, confirming that CSC are relatively inert to drug-induced apoptosis (Fig. 6a). This suggests that CSC might escape anti-tumour therapies and could be the underlying reason for chemotherapy inefficiency. 30 To formally prove that IL-4 production in colon CSC is responsible for up regulation of anti-apoptotic proteins and therefore therapy refractoriness, CSC were pre-treated for two days with IL-4-neutralising antibodies and then measured cell death and anti-apoptotic expression. Proteins levels of c- WO 2007/147600 PCT/EP2007/005480 19 FLIP, Bcl-xL and PED, anti-apoptotic proteins previously shown to be regulated by IL-4 in cancer, decreased by -two-fold in CSC exposed to anti IL-4 (Fig. 6b-c). More important, following IL-4 blockade CSC cell death was significantly increased by the treatment with chemotherapeutic drugs or 5 TRAIL (Fig. 6d-e). To directly demonstrate that IL-4 protects colon cancer generated by CSC from chemotherapeutic drugs, the effects of IL-4 neutralization in vivo were investigated. Tumours were allowed to grow for 10 days (size -0.2 cm 3 ) and 10 then treated intra-tumourally with neutralising antibodies against IL-4 or control IgG twice a week for 3 weeks. Although intraperitoneal (i.p.) treatment with oxaliplatin, once a week for 4 weeks, combined with control IgG reduced tumour size in mice, the efficacy of chemotherapy treatment was significantly enhanced by IL-4 neutralizing antibody (Fig. 7a and 7b). 15 Example 3 Construction of an IL4RIL13R-Fc fusion polypeptide 20 The signal-peptide and the extracellular domain of IL4-Receptor-alpha (aal aa231 of NCBI accession NP_000409) was fused N-terminally to the IL13 receptor alpha extracellular domain (aa27-aa343 of NCBI accession NP_001551) Two point mutations were introduced into the IL4R-alphal sequence (Gly2->Val2 and Cys207->Ser2O7) and a single point mutation 25 was introduced into the IL13R-alphal-sequence (Cys46->Ala46). The enumeration of the point mutations also refers to NCBI-database entries NP_000409 for IL4R-alphal and NP_001551 for IL13R-alphal. This IL4RIL13R protein-sequence was fused to the Fc-part of human IGHG1 30 (aa254-aa479 of NCBI accession AAH69020). Additionally, a flexible linker element and a Flexstreptag-ll motif (SSSSSSAWSHPQFEK) was added C terminally. The amino acid sequence of the resulting IL4RIL13R-Fc construct as shown below was backtranslated into a synthetic DNA- WO 2007/147600 PCT/EP2007/005480 20 sequence and its codon usage optimised for mammalian cell-based expression. Gene synthesis was done by ENTELECHON GmbH (Regensburg, Germany). The final expression cassette was subcloned into pCDNA4-HisMax-backbone, using the unique Hind-Ill- and Not-l-sites of the 5 plasmid. SEQ ID NO: 1 SEQ IL4RIL13R-Fc.PRO KEYWORD PROTEIN 10 ORIGIN 1 MVWLCSGLLF PVSCLVLLQV ASSGNMKVLQ EPTCVSDYMS ISTCEWKMNG PTNCSTELRL 61 LYQLVFLLSE AHTCIPENNG GAGCVCHLLM DDVVSADNYT LDLWAGQQLL WKGSFKPSEH 15 121 VKPRAPGNLT VHTNVSDTLL LTWSNPYPPD NYLYNHLTYA VNIWSENDPA DFRIYNVTYL 181 EPSLRIAAST LKSGISYRAR VRAWAQSYNT TWSEWSPSTK WHNSYREPFE QAPTETQPPV 241 TNLSVSVENL ATVIWTWNPP EGASSNCSLW YFSHFGDKQD KKIAPETRRS 20 IEVPLNERIC 301 LQVGSQCSTN ESEKPSILVE KCISPPEGDP ESAVTELQCI WHNLSYMKCS WLPGRNTSPD 361 TNYTLYYWHR SLEKIHQCEN IFREGQYFGC SFDLTKVKDS SFEQHSVQIM VKDNAGKIKP 25 421 SFNIVPLTSR VKPDPPHIKN LSFHNDDLYV QWENPQNFIS RCLFYEVEVN NSQTETHNVF 481 YVQEAKCENP EFERNVENTS CFMVPGVLPD TLNTVRIRVK TNKLCYEDDK LWSNWSQEMS 541 IGKKRNSTGD KTHTCPPCPA PELLGGPSVF LFPPKPKDTL MISRTPEVTC 30 VVVDVSHEDP 601 EVKFNWYVDG VEVHNAKTKP REEQYNSTYR VVSVLTVLHQ DWLNGKEYKC KVYNKALPAP 661 IEKTISKAKG QPREPQVYTL PPSREEMTKN QVSLTCLVKG FYPSDIAVEW ESNGQPENNY 35 721 KTTPLVLDSD GSFFLYSKLT VDKSRWQQGN VFSCSVMHEA LHNHYTQKSL SLSPGSSSSS 781 SAWSHPQFEK aal-aa23: signal peptide 40 aa24-aa231: IL4R-alphal ECD aa232-aa548: IL13R-alphal ECD aa549-aa775: Fc part of IGHG1 aa786-aa790: Flexstreptag-II 45 Modifications of the IL4R-IL1 3R-Fc fusion polypeptide may be as follows: - absence of the signal peptide or presence of a heterologous signal peptide; - presence of a different, e.g. shortened IL-4R ECD, e.g. without or with WO 2007/147600 PCT/EP2007/005480 21 different mutations, particularly point mutations, - presence of a different effector domain, - presence of a different Fc domain, and/or - absence of the C-terminal purification domain (particularly for 5 pharmaceutical applications). Example 4 Construction of an IL4R-Fc fusion polypeptide 10 The signal-peptide and a shortened extracellular domain of IL4-Receptor alpha (aal-aa224 of NCBI accession NP_000409) was fused N-terminally to the Fc-part of human IGHG1 (aa250-aa479 of NCBI accession AAH69020). Two point mutations were introduced into the IL4R-alphal-sequence (Gly2 15 >Val2 and Cys207->Ser207). A single glycine was inserted inbetween the two domains and Lys251 of human IGHG1 in the hinge region was mutated to arginine. The enumeration of the described mutations also refer to NCBI database entries NP_000409 for IL4R-alphal and NCBI accession AAH69020 for IGHG1). 20 Additionally, a flexible linker element and a Flexstreptag-II motif (SSSSSSAWSHPQFEK) was added C-terminally. The amino acid sequence of the resulting IL4R-Fc-construct as shown below was backtranslated into a synthetic DNA-sequence and its codon usage optimised for mammalian cell 25 based expression. Gene synthesis was done by ENTELECHON GmbH (Regensburg, Germany). The final expression cassette was subcloned into pCDNA4-HisMax-backbone, using the unique Hind-Ill- and Not-l-sites of the plasmid. 30 SEQ ID NO:2 SEQ IL4RA-Fc.PRO KEYWORD PROTEIN COLOURS 35 sequence = 1 WO 2007/147600 PCT/EP2007/005480 22 features = 0 ORIGIN 1 MVWLCSGLLF PVSCLVLLQV ASSGNMKVLQ EPTCVSDYMS ISTCEWKMNG PTNCSTELRL 5 61 LYQLVFLLSE AHTCIPENNG GAGCVCHLLM DDVVSADNYT LDLWAGQQLL WKGSFKPSEH 121 VKPRAPGNLT VHTNVSDTLL LTWSNPYPPD NYLYNHLTYA VNIWSENDPA DFRIYNVTYL 181 EPSLRIAAST LKSGISYRAR VRAWAQSYNT TWSEWSPSTK WHNSGSRSCD 10 KTHTCPPCPA 241 PELLGGPSVF LFPPKPKDTL MISRTPEVTC VVVDVSHEDP EVKFNWYVDG VEVHNAKTKP 301 REEQYNSTYR VVSVLTVLHQ DWLNGKEYKC KVSNKALPAP IEKTISKAKG QPREPQVYTL 15 361 PPSREEMTKN QVSLTCLVKG FYPSDIAVEW ESNGQPENNY KTTPPVLDSD GSFFLYSKLT 421 VDKSRWQQGN VFSCSVMHEA LHNHYTQKSL SLSPGSSSSS SAWSHPQFEK Aal-aa23: IL4R-alphal signal peptide 20 Aa24-aa224: IL4R-alphal ECD Aa225-aa455: Fc part of IGHG1 Aa456-aa470: Flexstreptag-II Modifications of the shortened IL-4R fusion polypeptide may be as follows: 25 - absence of a signal peptide or presence of a heterologous signal peptide; - presence of the different, e.g. shortened lL-4R ECD, e.g. without or with different mutations, particularly point mutations, - presence of a different Fc domain, 30 - a different fusion region between the IL-4R ECD and the Fc domain, e.g. deletion of one or more amino acids of the sequence RSC (positions 227-229), and/or - absence of the C-terminal purification domain (particularly for pharmaceutical applications). 35 Example 5 Expression and Purification of lL4-binding proteins, IL4R-Fc and IL4R IL1 3R-Fc 40 Hek 293T cells grown in DMEM + GlutaMAX (GibCo) supplemented with 10 % FBS, 100 units/ml Penicillin and 100 pg/ml Streptomycin were transiently WO 2007/147600 PCT/EP2007/005480 23 transfected with plasmids encoding IL4R-Fc and IL4R-IL13R-Fc, respectively. Cell culture supernatants containing recombinant proteins were harvested three days post transfection and clarified by centrifugation at 300 g followed by filtration through a 0.22 pm sterile filter. For affinity 5 purification Streptactin Sepharose was packed to a column (gel bed 1 ml), equilibrated with 15 ml buffer W (100 mM Tris-HCl, 150 mM NaCl pH 8.0) and the respective cell culture supernatant was applied to the column with a flow rate of 4 ml/min. Subsequently, the column was washed with buffer W and bound IL4R-Fc or IL4R-IL13R-Fc was eluted stepwise by addition of 6 x 10 1 ml buffer E (100 mM Tris HCI, 150 mM NaCl, 2.5 mM Desthiobiotin pH 8.0). The protein amount of the eluate fractions was quantified and peak fractions were concentrated by ultrafiltration and further purified by size exclusion chromatography (SEC). An SDS-PAGE of the Streptactin affinity purification of IL4R-IL13R-Fc followed by Silver staining is shown in Figure 1s 8A. SEC was performed on a Superdex 200 column using an Akta chromatography system (GE-Healthcare). The column was equilibrated with phosphate buffered saline and the concentrated, streptactin purified IL4R 20 Fc or IL4R-IL13R-Fc, respectively, were loaded onto the SEC column at a flow rate of 0.5 ml/min. The elution profile monitored by absorbance at 280 nm showed a prominent protein peak at 10.31 ml for IL4R-IL13R-Fc (Figure 8B ) and 12.97 ml for IL4R-Fc (Figure 9A). SEC fractions for IL4R-Fc were additionally analysed under denaturing conditions by SDS-PAGE and 25 silver staining (Figure 9B). For determination of the apparent molecular weight under native conditions a Superdex 200 column was loaded with standard proteins of known molecular weight. Based on the elution volume of the standard proteins a 30 calibration curve was calculated and the apparent molecular weight of purified IL4R-Fc was determined to be 137 KDa which fits well to the molecular weight observed by SDS-PAGE. The theoretical molecular weight based on the amino acid sequence of IL4R-Fc is 52.8 Kda for the WO 2007/147600 PCT/EP2007/005480 24 monomeric protein. Based on the biochemical analysis IL4R-Fc very likely is expressed as a protein dimer. For IL4R-IL13R-Fc the apparent molecular weight based on SEC was 5 calculated to be about 600 KDa. Based on SDS-Page analysis the protein runs as a single band with about 250Kda. The theoretical molecular weight based on the amino acid sequence of IL4R-IL13R-Fc is 87.7 KDa. In principle the construction of the molecule should result in a stable dimeric protein with a theoretical molecular weight of about 180Kda. The high 10 apparent molecular weight seen by SEC therefore either indicates an unusual behavior in SEC or further oligomerisation of the protein. IL4-pull down assay 15 To test for specific 1L4 binding of IL4R-Fc and IL4R-IL13R-Fc, 4pg of both proteins, respectively, were immobilized to Streptactin Sepharose via their Strep-Tag. The immobilized proteins were subsequently incubated for 60 min with 400 ng of recombinantly expressed human Interleukin4 (1L4) in a total volume of 400 pl phosphate buffered saline. Subsequently the beads 20 were washed and bound proteins were specifically eluted with desthiobiotin in a total volume of 40 pl elution buffer. Eluted proteins were finally analysed via SDS-PAGE and Silver staining. As shown in Figure 10 both IL4R-Fc and IL4R-IL13R-Fc show specific binding of human 1L4 indicated by the presence of 1L4 protein (12Kda) that could not be seen in control 25 experiments. Example 6 In vitro efficacy on Cancer stem cells and primary tumor cells 30 To test for the ability of IL4R-Fc and IL4R-IL13R-Fc to induce apoptosis, both proteins were added to the growth medium of breast cancer stem cells either alone or in combination with doxorubicin. Figure 11A shows the WO 2007/147600 PCT/EP2007/005480 25 immunofluorescence analysis of breast cancer spheres pre-treated with PBS (w/o) or 10 pg of IL4R-Fc, IL4R-IL13R-Fc or anti IL4-antibody for 24 hrs and successively exposed for another 24 hrs to 5 pM doxorubicin. The cells were stained with orange acridine/ethidium bromide (red: dead cells; green: viable 5 cells). In comparison with the single treatment (Doxorubicin alone) the combination of doxorubicin with either IL4R-Fc or IL4R-IL13R-Fc, respectively, clearly increased the amout of apoptotic breast cancer stem cells. A cell count discriminating apoptotic and living cells, subsequently plotted for the percentage of cell viability also demonstrates the efficacy of 10 the combination treatmant for the induction of apoptosis (Figure 11 B). Importantly both IL4R-Fc and IL4R-IL13R-Fc are able to sensitise breast cancer stem cells for doxorubicin induced apoptosis in the same range as shown for an IL4 specific antibody, that was used as a positive control in this experiment. 15 On primary colon cancer cells the IL4R-Fc and IL4R-IL13R-Fc constructs were tested in combination with oxaliplatin treatment. Primary colon cancer cells pre-treated with PBS (w/o) or 10 pg of IL4R-Fc, IL4R-IL13R-Fc or anti IL4-antibody for 24 hrs and successively exposed for another 24 hrs to 20 100 pM oxaliplatin. The graphs show the percentage of cell viability measured by MTS analysis (CellTiter 96, Aquos, Promega ). As shown in Figure 11 C, both constructs are able to sensitze primary colon cancer cells for oxaliplatin induced apoptosis, indicated by a reduced cell viability in comparison with oxaliplatin treatment alone. 25

Claims (24)

  1. 2. The method according to claim 1 wherein the anti-apoptotic cytokine is IL-4 and/or IL-10, preferably IL-4. 15 3. The method according to claims 1 and 2, wherein the solid tumour is classified as an IL-4 expressing or an IL-4 non-expressing tumour.
  2. 4. The method according to claims 1 and 2, wherein the solid tumour is classified as an IL-10 expressing or an IL-10 non-expressing tumour.
  3. 5. The method according to claims 1 to 4 wherein the solid tumour is classified as an IL-4 and IL-10 expressing tumour or as a non-IL-4 and a non-IL-10 expressing tumour. 25 6. The method according to any of the preceding claims, wherein the solid tumour is an epithelial tumour.
  4. 7. The method according to claim 6, wherein the epithelial tumour is selected from the group of thyroid, breast, prostate, bladder, colon, 30 gastric, pancreas, kidney, liver and lung cancer.
  5. 8. The method according to claim 7 wherein the tumour preferably is a colon, gastric, breast, lung, bladder, or prostate cancer. WO 2007/147600 PCT/EP2007/005480 27
  6. 9. The method according to any of the preceding claims, wherein the tumour cells are primary tumour cells and/or cancer stem cells.
  7. 10. The method according to any of the preceding claims, wherein detecting 5 the anti-apoptotic cytokine expression in the tumour cells comprises a detection on the protein level and/or on the nucleic acid level.
  8. 11. The method according to claim 10 wherein the detection on the protein level comprises the detection of the anti-apoptotic cytokine, preferably 10 with immunochemical and /or mass spectrometric methods.
  9. 12. The method according to claim 10, wherein the determination on nucleic acid level comprises the determination of anti-apoptotic cytokine mRNA expression levels with nucleic acid hybridization and optionally 15 amplification methods, preferably with RT-PCR methods.
  10. 13. The method according to any of the preceding claims, further comprising the steps of (d) determining the sensitivity of the cells of a cytokine expressing 20 tumour against at least one chemotherapeutic or pro-apoptotic agent in the presence and/or in the absence of an antagonist of said expressed cytokine, and/or its receptor and (e) optionally selecting a cancer type-specific treatment. 25 14. The method of claim 13 wherein in step (d) a chemotherapeutic or pro apoptotic agent is determined against which the cells of the cytokine expressing tumour are sensitive.
  11. 15. The method according to claim 13 or 14, wherein, in step 13(e), a 30 treatment is selected comprising the administration of a combination of a cytokine neutralizing agent and a chemotherapeutic or pro-apoptotic agent. WO 2007/147600 PCT/EP2007/005480 28
  12. 16. The method according to claim 14 or 15 wherein the chemotherapeutic agent is selected from antimetabolites, DNA-fragmenting agents, DNA crosslinking agents, intercalating agents, protein synthesis inhibitors, topoisomerase I and 11 inhibitors, microtubule-directed agents, kinase 5 inhibitors, hormones and hormone antagonists.
  13. 17. The method according to claim 16 wherein the chemotherapeutic agent is selected from cisplatin, carboplatin and oxaliplatin. 10 18. The method according to claim 14 or 15, wherein the pro-apoptotic agent is selected from TRAIL and CD95 ligand.
  14. 19. The method according to any one of claims 14 to 18, wherein the cytokine neutralizing agent is an antibody, preferably an anti-IL-4 15 antibody and/or an anti-IL-10 antibody or an antigen-binding fragment thereof.
  15. 20. The method according to claim 19, wherein the anti-IL-4 antibody is an antibody derived from the hybridoma cell ECACC 93100620 or an 20 antigen-binding fragment thereof.
  16. 21. The method according to any of claims 14 to 18, wherein the cytokine neutralizing agent is a soluble IL-4 receptor polypeptide or fusion polypeptide.
  17. 22. The use of a combination of (i) at least one cytokine neutralizing agent and (ii) at least a chemotherapeutic or pro-apoptotic agent for the manufacture of a medicament for the treatment of minimal 30 residual disease.
  18. 23. The use of a combination of (i) at least one cytokine neutralizing agent and WO 2007/147600 PCT/EP2007/005480 29 (ii) at least a chemotherapeutic or pro-apoptotic agent for the manufacture of a medicament for the treatment of a cancer type classified as cytokine-expressing tumour. 5 24. The use of a combination of (i) at least one cytokine neutralizing agent and (ii) at least a chemotherapeutic or pro-apoptotic agent for the manufacture of a medicament for the treatment of a cancer type classified as cytokine-expressing tumour in combination with surgery 10 and/or irradiation therapy.
  19. 25. The use according to claim 24, wherein the medicament is for simultaneous, separate or sequential combination therapy with surgery and/or irradiation therapy.
  20. 26. The use of a combination of (i) at least one cytokine neutralizing agent and (ii) at least a chemotherapeutic or pro-apoptotic agent for the manufacture of a medicament for the treatment of a cytokine 20 expressing tumour wherein the administration of (i) and (ii) is started simultaneously.
  21. 27. The use of a combination of (i) at least one cytokine neutralizing agent and 25 (ii) at least a chemotherapeutic or pro-apoptotic agent for the manufacture of a medicament for the treatment of a cytokine expressing tumour wherein the administration of (i) and (ii) is started stepwise. 30 28. The use according to claim 27 wherein the start of administration of (i) is 2 1 week before (ii) or wherein the start of administration of (ii) is 2 1 week before (i). WO 2007/147600 PCT/EP2007/005480 30
  22. 29. A soluble IL-4 receptor polypeptide comprising a C-terminally shortened extracellular IL-4 receptor domain.
  23. 30. The polypeptide of claim 29 which is a fusion polypeptide.
  24. 31. A nucleic acid molecule encoding the polypeptide of claim 29 or 30.
AU2007263265A 2006-06-21 2007-06-21 Differential IL-4 and/or IL-10 cytokine expression in human cancer Abandoned AU2007263265A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP06012754 2006-06-21
EP06012754.5 2006-06-21
PCT/EP2007/005480 WO2007147600A2 (en) 2006-06-21 2007-06-21 Differential il-4 and/or il-10 cytokine expression in human cancer

Publications (1)

Publication Number Publication Date
AU2007263265A1 true AU2007263265A1 (en) 2007-12-27

Family

ID=38359749

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2007263265A Abandoned AU2007263265A1 (en) 2006-06-21 2007-06-21 Differential IL-4 and/or IL-10 cytokine expression in human cancer

Country Status (12)

Country Link
US (1) US20090324616A1 (en)
EP (1) EP2041576B1 (en)
JP (1) JP2010514409A (en)
CN (1) CN101529253A (en)
AT (1) ATE520032T1 (en)
AU (1) AU2007263265A1 (en)
BR (1) BRPI0713484A2 (en)
CA (1) CA2656379A1 (en)
DK (1) DK2041576T3 (en)
ES (1) ES2371287T3 (en)
RU (1) RU2009101783A (en)
WO (1) WO2007147600A2 (en)

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007107349A1 (en) * 2006-03-22 2007-09-27 Apogenix Gmbh Antibody specific for human il-4 for the treament of cancer
CA2656135A1 (en) * 2006-07-06 2008-01-10 Apogenix Gmbh Human il-4 muteins in cancer therapy
JP6231562B2 (en) * 2012-07-18 2017-11-15 アポジェニックス アーゲー Composition comprising a mixture of CD95-Fc isoforms
US11299528B2 (en) 2014-03-11 2022-04-12 D&D Pharmatech Inc. Long acting TRAIL receptor agonists for treatment of autoimmune diseases
BR112017008399A2 (en) * 2014-10-24 2018-06-19 StemImmune, Incorporated Combination immunotherapy approach for cancer treatment
CA3004891C (en) 2015-08-11 2020-04-14 StemImmune, Incorporated Smallpox vaccine for cancer treatment
CN108601819B (en) 2015-12-17 2022-03-15 约翰霍普金斯大学 Amelioration of systemic sclerosis with death receptor agonists
EA201892260A1 (en) 2016-04-07 2019-03-29 Дзе Джонс Хопкинс Юниверсити COMPOSITIONS AND METHODS FOR THE TREATMENT OF PANCREATITIS AND PAIN WITH THE APPLICATION OF THE AGONISTS OF THE DEATH RECEPTOR
US20190317098A1 (en) * 2016-11-24 2019-10-17 Huvet Bio, Inc. Composition for diagnosis of diseases
CA3071661A1 (en) * 2017-09-06 2019-03-14 Fred Hutchinson Cancer Research Center Strep-tag specific chimeric receptors and uses thereof
US11505782B2 (en) 2018-06-04 2022-11-22 Calidi Biotherapeutics, Inc. Cell-based vehicles for potentiation of viral therapy

Family Cites Families (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU643427B2 (en) * 1988-10-31 1993-11-18 Immunex Corporation Interleukin-4 receptors
DE4137333A1 (en) * 1991-11-13 1993-05-19 W Prof Dr Sebald THERAPEUTIC AGENTS THAT ARE ANTAGONISTS OR PARTIAL AGONISTS OF THE HUMAN INTERLEUKIN 4 OR CONTAIN THEM, HIL-4-MUTANT PROTEINS AND METHOD FOR THE PRODUCTION THEREOF
US6534051B1 (en) * 1992-11-20 2003-03-18 University Of Medicine And Dentistry Of New Jersey Cell type specific gene transfer using retroviral vectors containing antibody-envelope fusion proteins and wild-type envelope fusion proteins
ES2236693T3 (en) 1993-09-07 2005-07-16 Smithkline Beecham Corporation RECOMBINANT ANTIBODIES AGAINST IL4 USEFUL IN THE TREATMENT OF CONDITIONS AFTER IL4.
DE4423131A1 (en) * 1994-07-01 1996-01-04 Bayer Ag New hIL-4 mutant proteins as antagonists or partial agonists of human interleukin 4
WO1996001318A1 (en) * 1994-07-05 1996-01-18 Steeno Research Group A/S Immunomodulators
CN1117155C (en) * 1994-07-29 2003-08-06 史密丝克莱恩比彻姆有限公司 Novel compounds
US5710023A (en) * 1996-03-01 1998-01-20 Genetics Institute, Inc. IL-13 cytokine receptor chain
US6664227B1 (en) * 1996-03-01 2003-12-16 Genetics Institute, Llc Treatment of fibrosis by antagonism of IL-13 and IL-13 receptor chains
US20030124125A1 (en) * 1996-04-05 2003-07-03 South Alabama Medical Science Foundation Oncofetal antigen specific T-lymphocyte mediated immune response: manipulation and uses of oncofetal antigen specific CD4, CD8 cytotoxic and suppressor T cells and interleukin-10
US6028176A (en) * 1996-07-19 2000-02-22 Bayer Corporation High-affinity interleukin-4 muteins
CA2309598A1 (en) * 1997-11-10 1999-05-20 Mochida Pharmaceutical Co., Ltd. Preventives and remedies for diffuse lung disease
JPH11312463A (en) * 1998-04-28 1999-11-09 Hitachi Ltd Wiring board and gas discharge display device using it
JP2004500412A (en) * 2000-03-31 2004-01-08 アイデック ファーマスーティカルズ コーポレイション Combination of anti-cytokine antibody or antagonist and anti-CD20 for treatment of B-cell lymphoma
WO2002004009A2 (en) * 2000-07-12 2002-01-17 Immunex Corporation Method for treating cancer using an interleukin- 4 antagonist
US20040023338A1 (en) * 2001-10-26 2004-02-05 Heavner George A. IL-4 mutein proteins, antibodies, compositions, methods and uses
WO2003035847A2 (en) * 2001-10-26 2003-05-01 Centocor, Inc. Il-13 mutein proteins, antibodies, compositions, methods and uses
EP1444989A1 (en) 2003-02-07 2004-08-11 Giorgio Dr. Stassi Sensitizing cells for apoptosis by selectively blocking cytokines
AR049390A1 (en) * 2004-06-09 2006-07-26 Wyeth Corp ANTIBODIES AGAINST HUMAN INTERLEUQUINE-13 AND USES OF THE SAME
US7501121B2 (en) * 2004-06-17 2009-03-10 Wyeth IL-13 binding agents
CA2587903A1 (en) * 2004-11-17 2006-05-26 Amgen Fremont Inc. Fully human monoclonal antibodies to il-13
US20070122855A1 (en) * 2005-11-28 2007-05-31 Targetgen Inc. Methods for diagnosing hepatocellular carcinoma
WO2007107349A1 (en) * 2006-03-22 2007-09-27 Apogenix Gmbh Antibody specific for human il-4 for the treament of cancer
CA2656135A1 (en) * 2006-07-06 2008-01-10 Apogenix Gmbh Human il-4 muteins in cancer therapy

Also Published As

Publication number Publication date
EP2041576B1 (en) 2011-08-10
ATE520032T1 (en) 2011-08-15
ES2371287T3 (en) 2011-12-29
RU2009101783A (en) 2010-07-27
DK2041576T3 (en) 2011-12-05
EP2041576A2 (en) 2009-04-01
BRPI0713484A2 (en) 2012-11-06
CN101529253A (en) 2009-09-09
WO2007147600A2 (en) 2007-12-27
WO2007147600A3 (en) 2008-04-10
CA2656379A1 (en) 2007-12-27
JP2010514409A (en) 2010-05-06
US20090324616A1 (en) 2009-12-31

Similar Documents

Publication Publication Date Title
EP2041576B1 (en) Differential cytokine expression in human cancer
Du et al. The co-expression characteristics of LAG3 and PD-1 on the T cells of patients with breast cancer reveal a new therapeutic strategy
JP6812364B2 (en) Anti-GITR antibody for cancer diagnosis
Todaro et al. Colon cancer stem cells dictate tumor growth and resist cell death by production of interleukin-4
DeFazio-Eli et al. Quantitative assays for the measurement of HER1-HER2 heterodimerization and phosphorylation in cell lines and breast tumors: applications for diagnostics and targeted drug mechanism of action
DK2533047T3 (en) CCR4 as a therapeutic target for cancer
Meinhold-Heerlein et al. Expression and potential role of Fas-associated phosphatase-1 in ovarian cancer
JP2014505239A (en) Use of anti-CXCL13 and anti-CXCR5 antibodies for the treatment or detection of cancer
Xie et al. Formyl peptide receptor 2 expression predicts poor prognosis and promotes invasion and metastasis in epithelial ovarian cancer
Costes et al. Expression of the interleukin 6 receptor in primary renal cell carcinoma.
Ekshyyan et al. Rapamycin targets Interleukin 6 (IL-6) expression and suppresses endothelial cell invasion stimulated by tumor cells
Liu et al. Desensitization of T lymphocyte function by CXCR3 ligands in human hepatocellular carcinoma
Castellani et al. Interaction of transforming growth factor‐alpha and epidermal growth factor receptor in breast carcinoma. An immunohistologic study
CN107249636B (en) Antitumor agent containing CKAP4 as target molecule
KR20210107730A (en) Use of IL-1 beta antibodies in the treatment or prevention of myelodysplastic syndrome
DK2585089T3 (en) CCL18 blockade of signaling through CCR6 as a therapeutic option in fibrotic diseases and cancer
US8969020B2 (en) Peptide sequence that promotes tumor invasion
WO2010066858A1 (en) Methods for the treatment and the prognosis of cancer
US20120015352A1 (en) Method of determining sensitivity of human or non-human animal cells to an iap antagonist
Wu Circulating tumor markers of the new millennium: target therapy, early detection, and prognosis
JP2006507243A (en) Proteins related to cancer
Zhu et al. Expression of chemokines CCL5 and CCL11 by smooth muscle tumor cells of the uterus and its possible role in the recruitment of mast cells
CA3076997C (en) Anti-ckap4 monoclonal antibody
Seol et al. Aggressive vestibular schwannomas showing postoperative rapid growth–their association with decreased p27 expression
KR102268041B1 (en) A Composition for predicting the prognosis of oral carcinoma

Legal Events

Date Code Title Description
MK1 Application lapsed section 142(2)(a) - no request for examination in relevant period